Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
2.
Eur J Vasc Endovasc Surg ; 57(2): 259-266, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30343000

RESUMO

OBJECTIVES: One third of infrainguinal vein bypasses may fail within the first 1.5 years. Pro- and anti-inflammatory mechanisms are thought to be involved in these graft stenoses and occlusions. In previous studies, low levels of anti-phosphorylcholine IgM (anti-PC IgM, an innate anti-inflammatory IgM) have been associated with increased cardiovascular events. In this study, the peri-operative dynamics of anti-PC IgM levels were established during leg bypass surgery, and associations assessed between anti-PC IgM levels and primary graft patency. DESIGN AND METHODS: This was a prospective, observational cohort study of infrainguinal autogenous vein bypass for peripheral arterial occlusive disease involving four university affiliated hospitals. Plasma cytokine and anti-PC IgM levels were measured pre- and post-operatively. The outcome of interest was loss of primary graft patency because of occlusion or intervention for graft stenosis. RESULTS: One hundred and forty-two consecutive patients were enrolled: mean age 66 (46-91); 91% white race and male; 72.5% critical limb ischaemia (Fontaine III or IV). Median pre-operative anti-PC IgM levels were 49 units/mL (IQR 32.3-107.7, mean 89.8 + 101 sd). During follow up of an average of 1.8 years (1 month-7.4 years), 50 (35.2%) grafts lost primary patency. Pre-operative levels of interleukin 6 or C-reactive protein did not predict graft failure. Patients with pre-operative anti-PC IgM values in the lowest quartile had a twofold increased risk of graft failure (multivariable Cox proportional hazard, p = .03, HR 2.11, 95% CI 1.09-4.07), even after accounting for the other significant factors of conduit diameter, distal anastomosis, smoking, and the severity of leg ischaemia. CONCLUSIONS: Low levels of anti-PC IgM are associated with vein bypass graft failure. This biological mediator may be a useful marker to identify patients at higher risk, and offers the potential for novel, directed therapies for vascular inflammation and its consequences.


Assuntos
Oclusão de Enxerto Vascular/cirurgia , Rejeição de Enxerto/diagnóstico , Imunoglobulina M/metabolismo , Doença Arterial Periférica/cirurgia , Fosforilcolina/imunologia , Enxerto Vascular/métodos , Idoso , Idoso de 80 Anos ou mais , Autoenxertos , Feminino , Oclusão de Enxerto Vascular/imunologia , Rejeição de Enxerto/imunologia , Humanos , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Doença Arterial Periférica/imunologia , Estudos Prospectivos , Veia Safena/cirurgia , Resultado do Tratamento , Grau de Desobstrução Vascular
3.
J Vasc Surg ; 68(6S): 165S-176S.e6, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29914830

RESUMO

OBJECTIVE: When an autogenous vein is harvested and used for arterial bypass, it suffers physical and biologic injuries that may set in motion the cellular processes that lead to wall thickening, fibrosis, stenosis, and ultimately graft failure. Whereas the injurious effects of surgical preparation of the vein conduit have been extensively studied, little is known about the influence of the clinical environment of the donor leg from which the vein is obtained. METHODS: We studied the cellular responses of fresh saphenous vein samples obtained before implantation in 46 patients undergoing elective lower extremity bypass surgery. Using an ex vivo model of response to injury, we quantified the outgrowth of cells from explants of the adventitial and medial layers of the vein. We correlated this cellular outgrowth with the clinical characteristics of the patients, including the Wound, Ischemia, and foot Infection classification of the donor leg for ischemia, wounds, and infection as well as smoking and diabetes. RESULTS: Cellular outgrowth was significantly faster and more robust from the adventitial layer than from the medial layer. The factors of leg ischemia (P < .001), smoking (P = .042), and leg infection (P = .045) were associated with impaired overall outgrowth from the adventitial tissue on multivariable analysis. Only ischemia (P = .046) was associated with impaired outgrowth of smooth muscle cells (SMCs) from the medial tissue. Co-culture of adventitial cells and SMCs propagated from vein explants revealed that adventitial cells significantly inhibited the growth of SMCs, whereas SMCs promoted the growth of adventitial cells. The AA genotype of the -838C>A p27 polymorphism (previously associated with superior graft patency) enhanced these effects, whereas the factor of smoking attenuated adventitial cell inhibition of SMC growth. Comparing gene expression, the cells cultured from the media overexpress Kyoto Encyclopedia of Genes and Genomes pathways associated with inflammation and infection, whereas those from the adventitia overexpress gene families associated with development and stem/progenitor cell maintenance. CONCLUSIONS: The adverse clinical environment of the leg may influence the biologic behavior of the cells in the vein wall, especially the adventitial cells. Chronic ischemia was the most significant factor that retards adventitial cell outgrowth. The cells arising from the vein adventitia may be key players in determining a healthy adaptive or a pathologic response to the injuries associated with vein grafting.


Assuntos
Isquemia/cirurgia , Extremidade Inferior/irrigação sanguínea , Doença Arterial Periférica/cirurgia , Veia Safena/transplante , Coleta de Tecidos e Órgãos/métodos , Enxerto Vascular/métodos , Idoso , Autoenxertos , Proliferação de Células , Células Cultivadas , Microambiente Celular , Técnicas de Cocultura , Inibidor de Quinase Dependente de Ciclina p27/genética , Feminino , Humanos , Isquemia/genética , Isquemia/metabolismo , Isquemia/patologia , Masculino , Pessoa de Meia-Idade , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Doença Arterial Periférica/genética , Doença Arterial Periférica/metabolismo , Doença Arterial Periférica/patologia , Polimorfismo Genético , Estudos Prospectivos , Fatores de Risco , Veia Safena/metabolismo , Veia Safena/patologia , Fumar/efeitos adversos , Fumar/metabolismo , Fumar/patologia , Técnicas de Cultura de Tecidos , Coleta de Tecidos e Órgãos/efeitos adversos , Enxerto Vascular/efeitos adversos , Grau de Desobstrução Vascular , Remodelação Vascular , Infecção dos Ferimentos/metabolismo , Infecção dos Ferimentos/patologia
4.
J Vasc Surg ; 67(5): 1556-1570.e9, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-28647196

RESUMO

OBJECTIVE: Venous valves are essential but are prone to injury, thrombosis, and fibrosis. We compared the behavior and gene expression of smooth muscle cells (SMCs) in the valve sinus vs nonvalve sites to elucidate biologic differences associated with vein valves. METHODS: Tissue explants of fresh human saphenous veins were prepared, and the migration of SMCs from explants of valve sinus vs nonvalve sinus areas was measured. Proliferation and death of SMCs were determined by staining for Ki67 and terminal deoxynucleotidyl transferase dUTP nick end labeling. Proliferation and migration of passaged valve vs nonvalve SMCs were determined by cell counts and using microchemotaxis chambers. Global gene expression in valve vs nonvalve intima-media was determined by RNA sequencing. RESULTS: Valve SMCs demonstrated greater proliferation in tissue explants compared with nonvalve SMCs (19.3% ± 5.4% vs 6.8% ± 2.0% Ki67-positive nuclei at 4 days, respectively; mean ± standard error of the mean, five veins; P < .05). This was also true for migration (18.2 ± 2.7 vs 7.5 ± 3.0 migrated SMCs/explant at 6 days, respectively; 24 veins, 15 explants/vein; P < .0001). Cell death was not different (39.6% ± 16.1% vs 41.5% ± 16.0% terminal deoxynucleotidyl transferase dUTP nick end labeling-positive cells, respectively, at 4 days, five veins). Cultured valve SMCs also proliferated faster than nonvalve SMCs in response to platelet-derived growth factor subunit BB (2.9 ± 0.2-fold vs 2.1 ± 0.2-fold of control, respectively; P < .001; n = 5 pairs of cells). This was also true for migration (6.5 ± 1.2-fold vs 4.4 ± 0.8-fold of control, respectively; P < .001; n = 7 pairs of cells). Blockade of fibroblast growth factor 2 (FGF2) inhibited the increased responses of valve SMCs but had no effect on nonvalve SMCs. Exogenous FGF2 increased migration of valve but not of nonvalve SMCs. Unlike in the isolated, cultured cells, blockade of FGF2 in the tissue explants did not block migration of valve or nonvalve SMCs from the explants. Thirty-seven genes were differentially expressed by valve compared with nonvalve intimal-medial tissue (11 veins). Peptide-mediated inhibition of SEMA3A, one of the differentially expressed genes, increased the number of migrated SMCs of valve but not of nonvalve explants. CONCLUSIONS: Valve compared with nonvalve SMCs have greater rates of migration and proliferation, which may in part explain the propensity for pathologic lesion formation in valves. Whereas FGF2 mediates these effects in cultured SMCs, the mediators of these stimulatory effects in the valve wall tissue remain unclear but may be among the differentially expressed genes discovered in this study. One of these genes, SEMA3A, mediates a valve-specific inhibitory effect on the injury response of valve SMCs.


Assuntos
Movimento Celular , Proliferação de Células , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Lesões do Sistema Vascular/patologia , Válvulas Venosas/patologia , Becaplermina , Morte Celular , Células Cultivadas , Fator 2 de Crescimento de Fibroblastos/farmacologia , Regulação da Expressão Gênica , Humanos , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Neointima , Proteínas Proto-Oncogênicas c-sis/farmacologia , Veia Safena/lesões , Veia Safena/metabolismo , Veia Safena/patologia , Semaforina-3A/genética , Semaforina-3A/metabolismo , Fatores de Tempo , Lesões do Sistema Vascular/genética , Lesões do Sistema Vascular/metabolismo , Válvulas Venosas/efeitos dos fármacos , Válvulas Venosas/lesões , Válvulas Venosas/metabolismo
5.
J Vasc Surg ; 67(1): 309-317.e7, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28526559

RESUMO

BACKGROUND: Cyclin-dependent kinase inhibitor 1B (p27Kip1) is a cell-cycle inhibitor whose -838C>A single nucleotide polymorphism (rs36228499; hereafter called p27 SNP) has been associated with the clinical failure of peripheral vein grafts, but the functional effects of this SNP have not been demonstrated. METHODS: Human saphenous vein adventitial cells and intimal/medial smooth muscle cells (SMCs) were derived from explants obtained at the time of lower extremity bypass operations. We determined the following in adventitial cells and SMCs as a function of the p27 SNP genotype: (1) p27 promoter activity, (2) p27 messenger (m)RNA and protein levels, and (3) growth and collagen gel contraction. Deoxyribonuclease I footprinting was also performed in adventitial cells and SMCs. RESULTS: p27 promoter activity, deoxyribonuclease I footprinting, p27 mRNA levels, and p27 protein levels demonstrated that the p27 SNP is functional in adventitial cells and SMCs. Both cell types with the graft failure protective AA genotype had more p27 mRNA and protein. As predicted because of higher levels of p27 protein, adventitial cells with the AA genotype grew slower than those of the CC genotype. Unexpectedly, SMCs did not show this genotype-dependent growth response. CONCLUSIONS: These results support the functionality of the p27 SNP in venous SMCs and adventitial cells, but an effect of the SNP on cell proliferation is limited to only adventitial cells. These data point to a potential role for adventitial cells in human vein graft failure and also suggest that SMCs express factors that interfere with the activity of p27.


Assuntos
Túnica Adventícia/fisiologia , Proliferação de Células/genética , Inibidor de Quinase Dependente de Ciclina p27/genética , Rejeição de Enxerto/genética , Miócitos de Músculo Liso/fisiologia , Veia Safena/transplante , Enxerto Vascular/efeitos adversos , Túnica Adventícia/citologia , Idoso , Células Cultivadas , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/metabolismo , Polimorfismo de Nucleotídeo Único , Cultura Primária de Células , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Veia Safena/citologia , Túnica Íntima/citologia , Túnica Íntima/fisiologia
6.
J Reprod Med ; 62(5-6): 329-32, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-30028098

RESUMO

OBJECTIVE: To determine if autologous endometrial cell coculture improves embryo development and clinical outcomes. STUDY DESIGN: Patients who met the inclusion criteria were randomized to either traditional in vitro fertilization (IVF) (control, n=73) or autologous endometrial cell coculture (AECC) (n=61). All patients underwent endometrial biopsy on cycle day 5­10 post luteinizing hormone surge. A total of 129 patients underwent embryo transfer (69 control, 60 AECC). Clinical outcomes as well as embryonic quality measures were then compared between the 2 groups. RESULTS: The mean age, day 3 follicle-stimulating hormone, number of oocytes collected, and clinical outcomes were similar between the 2 groups. Embryo development was overall similar, with the exception that embryonic grade was significantly better with AECC than with control: 1.5 (0.04) vs. 2.6 (0.03), p<0.0001. There was no difference in implantation, live birth or multiple gestation rates. CONCLUSION: This is one of the largest prospective randomized controlled trials of AECC versus traditional IVF. There was significant improvement in embryo morphology in the coculture group, although clinical outcomes were similar between the groups. Further studies are necessary to achieve enough power to fully delineate the effects of coculture on IVF outcome.


Assuntos
Técnicas de Cocultura , Endométrio , Fertilização in vitro , Transferência Embrionária , Endométrio/citologia , Feminino , Humanos , Gravidez , Estudos Prospectivos
8.
J Vasc Surg ; 64(1): 202-209.e6, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-25935274

RESUMO

OBJECTIVE: Approximately 30% of autogenous vein grafts develop luminal narrowing and fail because of intimal hyperplasia or negative remodeling. We previously found that vein graft cells from patients who later develop stenosis proliferate more in vitro in response to growth factors than cells from patients who maintain patent grafts. To discover novel determinants of vein graft outcome, we have analyzed gene expression profiles of these cells using a systems biology approach to cluster the genes into modules by their coexpression patterns and to correlate the results with growth data from our prior study and with new studies of migration and matrix remodeling. METHODS: RNA from 4-hour serum- or platelet-derived growth factor (PDGF)-BB-stimulated human saphenous vein cells obtained from the outer vein wall (20 cell lines) was used for microarray analysis of gene expression, followed by weighted gene coexpression network analysis. Cell migration in microchemotaxis chambers in response to PDGF-BB and cell-mediated collagen gel contraction in response to serum were also determined. Gene function was determined using short-interfering RNA to inhibit gene expression before subjecting cells to growth or collagen gel contraction assays. These cells were derived from samples of the vein grafts obtained at surgery, and the long-term fate of these bypass grafts was known. RESULTS: Neither migration nor cell-mediated collagen gel contraction showed a correlation with graft outcome. Although 1188 and 1340 genes were differentially expressed in response to treatment with serum and PDGF, respectively, no single gene was differentially expressed in cells isolated from patients whose grafts stenosed compared with those that remained patent. Network analysis revealed four unique groups of genes, which we term modules, associated with PDGF responses, and 20 unique modules associated with serum responses. The "yellow" and "skyblue" modules, from PDGF and serum analyses, respectively, correlated with later graft stenosis (P = .005 and P = .02, respectively). In response to PDGF, yellow was also associated with increased cell growth. For serum, skyblue was also associated with inhibition of collagen gel contraction. The hub genes for yellow and skyblue (ie, the gene most connected to other genes in the module), scavenger receptor class A member 5 (SCARA5) and suprabasin (SBSN), respectively, were tested for effects on proliferation and collagen contraction. Knockdown of SCARA5 increased proliferation by 29.9% ± 7.8% (P < .01), whereas knockdown of SBSN had no effect. Knockdown of SBSN increased collagen gel contraction by 24.2% ± 8.6% (P < .05), whereas knockdown of SCARA5 had no effect. CONCLUSIONS: Using weighted gene coexpression network analysis of cultured vein graft cell gene expression, we have discovered two small gene modules, which comprise 42 genes, that are associated with vein graft failure. Further experiments are needed to delineate the venous cells that express these genes in vivo and the roles these genes play in vein graft healing, starting with the module hub genes SCARA5 and SBSN, which have been shown to have modest effects on cell proliferation or collagen gel contraction.


Assuntos
Antígenos de Diferenciação/genética , Oclusão de Enxerto Vascular/genética , Proteínas de Neoplasias/genética , Receptores Depuradores Classe A/genética , Enxerto Vascular/efeitos adversos , Grau de Desobstrução Vascular/genética , Veias/transplante , Becaplermina , Linhagem Celular , Movimento Celular , Proliferação de Células , Análise por Conglomerados , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Predisposição Genética para Doença , Oclusão de Enxerto Vascular/diagnóstico , Oclusão de Enxerto Vascular/metabolismo , Oclusão de Enxerto Vascular/fisiopatologia , Humanos , Hiperplasia , Neointima , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Proteínas Proto-Oncogênicas c-sis/farmacologia , Interferência de RNA , Fatores de Risco , Biologia de Sistemas , Transfecção , Resultado do Tratamento , Veias/efeitos dos fármacos , Veias/metabolismo , Veias/fisiopatologia , Cicatrização
9.
J Vasc Surg ; 63(4): 1044-50, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25935273

RESUMO

OBJECTIVE: Markers containing dyes such as crystal violet (CAS 548-62-9) are routinely used on the adventitia of vein bypass grafts to avoid twisting during placement. Because little is known about how these dyes affect vein graft healing and function, we determined the effect of crystal violet on cell migration and proliferation, which are responses to injury after grafting. METHODS: Fresh human saphenous veins were obtained as residual specimens from leg bypass surgeries. Portions of the vein that had been surgically marked with crystal violet were analyzed separately from those that had no dye marking. In the laboratory, they were split into easily dissected inner and outer layers after removal of endothelium. This cleavage plane was within the circular muscle layer of the media. Cell migration from explants was measured daily as either (1) percentage of migration-positive explants, which exclusively measures migration, or (2) number of cells on the plastic surrounding each explant, which measures migration plus proliferation. Cell proliferation and apoptosis (Ki67 and TUNEL staining, respectively) were determined in dye-marked and unmarked areas of cultured vein rings. The dose-dependent effects of crystal violet were measured for cell migration from explants as well as for proliferation, migration, and death of cultured outer layer cells. Dye was extracted from explants with ethanol and quantified by spectrophotometry. RESULTS: There was significantly less cell migration from visibly blue compared with unstained outer layer explants by both methods. There was no significant difference in migration from inner layer explants adjacent to blue-stained or unstained sections of vein because dye did not penetrate to the inner layer. Ki67 staining of vein in organ culture, which is a measure of proliferation, progressively increased up to 6 days in nonblue outer layer and was abolished in the blue outer layer. Evidence of apoptosis (TUNEL staining) was present throughout the wall and not different in blue-stained and unstained vein wall segments. Blue outer layer explants had 65.9 ± 8.0 ng dye/explant compared with 2.1 ± 1.3 for nonblue outer layer explants. Dye applied in vitro to either outer or inner layer explants dose dependently inhibited migration (IC50∼10 ng/explant). The IC50s of crystal violet for outer layer cell proliferation and migration were 0.1 and 1.2 µg/mL, whereas the EC50 for death was between 1 and 10 µg/mL. CONCLUSIONS: Crystal violet inhibits venous cell migration and proliferation, indicating that alternative methods should be considered for marking vein grafts.


Assuntos
Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Corantes/toxicidade , Violeta Genciana/toxicidade , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Equipamentos Cirúrgicos , Cicatrização/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Biomarcadores/metabolismo , Relação Dose-Resposta a Droga , Desenho de Equipamento , Humanos , Antígeno Ki-67/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Músculo Liso Vascular/cirurgia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Técnicas de Cultura de Órgãos , Veia Safena/efeitos dos fármacos , Veia Safena/metabolismo , Veia Safena/patologia , Fatores de Tempo
10.
J R Stat Soc Series B Stat Methodol ; 77(2): 397-415, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25870521

RESUMO

We consider causal inference in randomized survival studies with right censored outcomes and all-or-nothing compliance, using semiparametric transformation models to estimate the distribution of survival times in treatment and control groups, conditional on covariates and latent compliance type. Estimands depending on these distributions, for example, the complier average causal effect (CACE), the complier effect on survival beyond time t, and the complier quantile effect are then considered. Maximum likelihood is used to estimate the parameters of the transformation models, using a specially designed expectation-maximization (EM) algorithm to overcome the computational difficulties created by the mixture structure of the problem and the infinite dimensional parameter in the transformation models. The estimators are shown to be consistent, asymptotically normal, and semiparametrically efficient. Inferential procedures for the causal parameters are developed. A simulation study is conducted to evaluate the finite sample performance of the estimated causal parameters. We also apply our methodology to a randomized study conducted by the Health Insurance Plan of Greater New York to assess the reduction in breast cancer mortality due to screening.

11.
J Vasc Surg ; 61(3): 630-5, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25499713

RESUMO

BACKGROUND: Accurate measurement of true aortic luminal diameter (ALD) is critical for endograft sizing in endovascular treatment of blunt thoracic aortic injury (BTAI), but ALD is dynamic and changes with respect to patients' hemodynamic status. This study aimed to characterize how ALD at the time of diagnosis of BTAI compares with ALD at the time of endovascular repair and later at follow-up. METHODS: This is an Institutional Review Board-approved, single-institution retrospective analysis of prospectively obtained data. Patients were included who presented between July 2007 and December 2012 with computed tomography angiography (CTA)-diagnosed BTAI; who underwent thoracic endovascular aortic repair (TEVAR); and who underwent preoperative CTA, intraoperative intravascular ultrasound (IVUS), and postimplantation CTA. Comparison measurements of the ALD were made among CTA and IVUS images at the level of the left subclavian artery (LSCA) and between initial CTA and postimplantation CTA at 10, 15, and 20 cm distal to the LSCA. Theoretical endograft sizes were determined and compared for each ALD at the LSCA. RESULTS: Twenty-two patients were included in the analysis. Mean age was 38 ± 14 years (range, 17-61 years), with 82% men and mean Injury Severity Score of 43 ± 11 (range, 24-66). Mean time from emergency department admission to initial CTA was -1.2 ± 5 hours (range, -13 to 11.5 hours; negative time implies imaging at an outside facility before admission). Mean time from initial CTA to IVUS was 1.2 ± 1.4 days (range, 2.5 hours-5.7 days) and from IVUS to postimplantation CTA 33 ± 45 days (range, 17 hours-169 days). Overall, ALD measured by IVUS was significantly larger than that by initial CTA (Δ2.5 ± 3.1 mm; P < .05). ALD was also larger at 10, 15, and 20 cm distal to the LSCA in comparing the postimplantation CTA with the initial CTA (Δ2.4, 2.0, and 2.0 mm, respectively; all P < .05). More than half the devices would be sized differently with ALD measured by IVUS at the time of TEVAR vs initial CTA. CONCLUSIONS: The ALD of patients with BTAI is significantly larger when it is measured by IVUS at the time of TEVAR compared with at the time of initial CTA. This difference in ALD may translate to undersizing of endografts used in TEVAR for BTAI. IVUS at the time of TEVAR provides a more accurate measurement of the actual ALD and should be used for endograft sizing for patients with BTAI.


Assuntos
Aorta Torácica/diagnóstico por imagem , Aorta Torácica/cirurgia , Implante de Prótese Vascular/instrumentação , Prótese Vascular , Procedimentos Endovasculares/instrumentação , Desenho de Prótese , Ultrassonografia de Intervenção , Lesões do Sistema Vascular/diagnóstico por imagem , Lesões do Sistema Vascular/cirurgia , Ferimentos não Penetrantes/diagnóstico por imagem , Ferimentos não Penetrantes/cirurgia , Adolescente , Adulto , Aorta Torácica/lesões , Aorta Torácica/fisiopatologia , Aortografia/métodos , Feminino , Hemodinâmica , Humanos , Escala de Gravidade do Ferimento , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Estudos Retrospectivos , Fatores de Tempo , Tempo para o Tratamento , Tomografia Computadorizada por Raios X , Resultado do Tratamento , Lesões do Sistema Vascular/fisiopatologia , Washington , Ferimentos não Penetrantes/fisiopatologia , Adulto Jovem
12.
J Vasc Surg ; 58(4): 997-1005.e1-2, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23856610

RESUMO

BACKGROUND: All humans have natural, protective antibodies directed against phosphorylcholine (PC) epitopes, a common inflammatory danger signal appearing at sites of cell injury, oxidative stress, and on bacterial capsules. In large human cohorts, low levels of anti-PC IgM were associated with a significantly increased risk of stroke or myocardial infarction. However, it is not known if these antibodies protect against the premature closure of arterial reconstructions. METHODS: A prospective, observational study of patients undergoing elective, infrainguinal, autogenous vein bypasses for atherosclerotic occlusive disease of the legs was conducted. Clinical data were recorded prospectively, and preoperative levels of anti-PC IgM measured with the CVDefine kit from Athera Biotechnologies (Solna, Sweden). The principal clinical end point was the loss of primary patency (loss of graft flow, or any intervention for stenosis). Patients were followed regularly by duplex ultrasound at 1, 3, 6, 12, 18 months, and yearly thereafter. RESULTS: Fifty-six patients were studied, for an average of 1.3 years. Indications for surgery were claudication (33.9%), ischemic rest pain (17.9%), and ischemia with ulceration or gangrene (48.2%). Seventeen (30.4%) patients experienced loss of primary patency (10 graft occlusions, seven surgical or endovascular revisions of graft stenoses). Kaplan-Meier survival analysis showed that the quartile of patients with the lowest anti-PC IgM levels had significantly worse primary graft patency (log-rank test, P = .0085). Uni- and multivariate Cox proportional hazards analysis revealed that the preoperative anti-PC IgM level was an important predictor of graft failure. Patients with IgM values in the lowest quartile had a 3.6-fold increased risk of graft failure (95% confidence interval: 1.1-12.1), even after accounting for other significant clinical or technical factors such as indication for surgery, site of distal anastomosis, or vein graft diameter. CONCLUSIONS: A naturally occurring IgM antibody directed against the proinflammatory epitope PC may be protective against vein graft stenosis and failure, through anti-inflammatory mechanisms. Measurement of this antibody may be a useful prognostic indicator, although larger studies of more diverse populations will be needed to confirm these results. The biological actions of anti-PC IgM suggest it may be useful in developing immunotherapies to improve bypass longevity.


Assuntos
Aterosclerose/cirurgia , Oclusão de Enxerto Vascular/imunologia , Imunoglobulina M/sangue , Extremidade Inferior/irrigação sanguínea , Fosforilcolina/imunologia , Veias/transplante , Idoso , Idoso de 80 Anos ou mais , Aterosclerose/sangue , Aterosclerose/imunologia , Biomarcadores/sangue , Constrição Patológica , Regulação para Baixo , Feminino , Oclusão de Enxerto Vascular/sangue , Oclusão de Enxerto Vascular/diagnóstico por imagem , Oclusão de Enxerto Vascular/fisiopatologia , Humanos , Estimativa de Kaplan-Meier , Modelos Lineares , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Modelos de Riscos Proporcionais , Estudos Prospectivos , Fatores de Risco , Fatores de Tempo , Falha de Tratamento , Ultrassonografia Doppler Dupla , Grau de Desobstrução Vascular , Veias/diagnóstico por imagem , Veias/imunologia , Veias/fisiopatologia
13.
J Vasc Surg ; 57(5): 1179-85.e1-2, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23312942

RESUMO

OBJECTIVE: Factors responsible for the variability in outcomes after lower extremity vein bypass grafting (LEVBG) are poorly understood. Recent evidence has suggested that a single nucleotide polymorphism (SNP) in the promoter region of the p27(Kip1) gene, a cell-cycle regulator, is associated with coronary in-stent restenosis. We hypothesized an association with vein graft patency. METHODS: This was a retrospective genetic association study nested within a prospective cohort of 204 patients from three referral centers undergoing LEVBG for claudication or critical ischemia. The main outcome measure was primary vein graft patency. RESULTS: All patients were followed up for a minimum of 1 year with duplex graft surveillance (median follow-up, 893 days; interquartile range, 539-1315). Genomic DNA was isolated and SNP analysis for the p27(Kip1)-838C>A variants was performed. Allele frequencies were correlated with graft outcome using survival analysis and Cox proportional hazards modeling. The p27(Kip1)-838C>A allele frequencies observed were CA, 53%; CC, 30%; and AA, 17%, satisfying Hardy-Weinberg equilibrium. Race (P = .025) and history of coronary artery disease (P = .027) were different across the genotypes; all other baseline variables were similar. Primary graft patency was greater among patients with the -838AA genotype (75% AA vs 55% CA/CC at 3 years; P = .029). In a Cox proportional hazards model including age, sex, race, diabetes, critical limb ischemia, redo (vs primary) bypass, vein type, and baseline C-reactive protein level, the p27(Kip1)-838AA genotype was significantly associated with higher graft patency (hazard ratio for failure, 0.4; 95% confidence interval, 0.17-0.93). Genotype was also associated with early (0-1 month) changes in graft lumen diameter by ultrasound imaging. CONCLUSIONS: These data suggest that the p27(Kip1)-838C>A SNP is associated with LEVBG patency and, together with previous reports, underscore a central role for p27(Kip1) in the generic response to vascular injury.


Assuntos
Inibidor de Quinase Dependente de Ciclina p27/genética , Oclusão de Enxerto Vascular/genética , Claudicação Intermitente/cirurgia , Isquemia/cirurgia , Extremidade Inferior/irrigação sanguínea , Doença Arterial Periférica/cirurgia , Polimorfismo de Nucleotídeo Único , Enxerto Vascular/efeitos adversos , Grau de Desobstrução Vascular/genética , Veias/transplante , Idoso , Estado Terminal , Feminino , Frequência do Gene , Estudos de Associação Genética , Predisposição Genética para Doença , Oclusão de Enxerto Vascular/diagnóstico por imagem , Oclusão de Enxerto Vascular/fisiopatologia , Humanos , Claudicação Intermitente/genética , Claudicação Intermitente/fisiopatologia , Isquemia/genética , Isquemia/fisiopatologia , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Doença Arterial Periférica/genética , Doença Arterial Periférica/fisiopatologia , Fenótipo , Regiões Promotoras Genéticas , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Fatores de Risco , Fatores de Tempo , Resultado do Tratamento , Ultrassonografia Doppler Dupla , Estados Unidos , Veias/diagnóstico por imagem , Veias/fisiopatologia
14.
Development ; 138(20): 4451-63, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21880786

RESUMO

Fibronectin (FN) is a major component of the extracellular matrix and functions in cell adhesion, cell spreading and cell migration. In the retina, FN is transiently expressed and assembled on astrocytes (ACs), which guide sprouting tip cells and deposit a provisional matrix for sprouting angiogenesis. The precise function of FN in retinal angiogenesis is largely unknown. Using genetic tools, we show that astrocytes are the major source of cellular FN during angiogenesis in the mouse retina. Deletion of astrocytic FN reduces radial endothelial migration during vascular plexus formation in a gene dose-dependent manner. This effect correlates with reduced VEGF receptor 2 and PI3K/AKT signalling, and can be mimicked by selectively inhibiting VEGF-A binding to FN through intraocular injection of blocking peptides. By contrast, AC-specific replacement of the integrin-binding RGD sequence with FN-RGE or endothelial deletion of itga5 shows little effect on migration and PI3K/AKT signalling, but impairs filopodial alignment along AC processes, suggesting that FN-integrin α5ß1 interaction is involved in filopodial adhesion to the astrocytic matrix. AC FN shares its VEGF-binding function and cell-surface distribution with heparan-sulfate (HS), and genetic deletion of both FN and HS together greatly enhances the migration defect, indicating a synergistic function of FN and HS in VEGF binding. We propose that in vivo the VEGF-binding properties of FN and HS promote directional tip cell migration, whereas FN integrin-binding functions to support filopodia adhesion to the astrocytic migration template.


Assuntos
Astrócitos/metabolismo , Fibronectinas/metabolismo , Integrinas/metabolismo , Neovascularização Fisiológica , Vasos Retinianos/crescimento & desenvolvimento , Vasos Retinianos/metabolismo , Animais , Movimento Celular , Matriz Extracelular/metabolismo , Fibronectinas/deficiência , Fibronectinas/genética , Heparitina Sulfato/metabolismo , Integrina alfa5beta1/química , Integrina alfa5beta1/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Oligopeptídeos/química , Fosfatidilinositol 3-Quinases/metabolismo , Domínios e Motivos de Interação entre Proteínas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Vasos Retinianos/inervação , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
15.
J Vasc Surg ; 54(4): 1124-30, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21906902

RESUMO

OBJECTIVE: Infrainguinal autogenous vein grafts are especially prone to narrowing and failure, and both inflammatory and thrombotic pathways are implicated. Platelets and monocytes are the key thrombo-inflammatory cells that arrive first at sites of vascular injury. These cells have potent interactions that recruit and activate one another, propagating thrombotic and inflammatory responses within the vessel wall. We therefore hypothesized that elevated levels of platelet-monocyte aggregates (PMA) might be associated with stenosis, and could possibly discriminate between patients with or without vein graft stenosis. METHODS: Thirty-six vascular surgery patients were studied, in a stable quiescent period after infrainguinal autogenous vein graft bypasses for occlusive disease. Eighteen patients had hemodynamically significant graft stenoses confirmed by imaging, and 18 were free from stenosis. The level of PMA in whole blood was quantified after blood draw using two-color flow cytometry. Three measurements were made per sample: the basal, in-vivo level of aggregates (baseline PMA); the predisposition to spontaneously generate PMA (spontaneous PMA); and PMA generation by the addition of exogenous thrombin receptor-activating peptide (stimulated PMA). The baseline, in-vivo level of PMA was estimated by immediate flow analysis. The predisposition to spontaneously generate PMA was measured after in vitro incubation. Responsiveness to thrombin stimulation of the blood was quantified by the in vitro dose response to an exogenous thrombin receptor-activating peptide (sfllrn). RESULTS: Baseline PMA levels were similar in patients with vein graft stenosis vs nonstenosis (14.8% ± 3.2 vs 10.1% ± 1.5, respectively, mean ± SEM). However, patients with stenosis showed higher spontaneous PMA levels (58.5% ± 4.5 vs 28.3% ± 4.3; P < .001) and higher stimulated PMA levels (P < .001; analysis of variance). Covariables of smoking, diabetes, statin, or antithrombotic therapy could not account for these differences. CONCLUSIONS: Platelet-monocyte reactivity may play a role in the development of vein graft stenoses. Those with/without stenosis differed primarily in their threshold, or predisposition to form aggregates (spontaneous PMA), while their basal circulating levels of PMA (baseline PMA) were similar. These measurements may unmask pathologic differences in thrombo-inflammatory responsiveness that are not apparent in basal measurements. Understanding the causes and mechanisms leading to abnormal platelet-monocyte responses may improve approaches to predicting or preventing vein graft stenosis.


Assuntos
Plaquetas/imunologia , Oclusão de Enxerto Vascular/imunologia , Monócitos/imunologia , Doença Arterial Periférica/cirurgia , Adesividade Plaquetária , Enxerto Vascular/efeitos adversos , Veias/transplante , Adulto , Idoso , Idoso de 80 Anos ou mais , Plaquetas/efeitos dos fármacos , Estudos de Casos e Controles , Distribuição de Qui-Quadrado , Constrição Patológica , Feminino , Citometria de Fluxo , Oclusão de Enxerto Vascular/sangue , Oclusão de Enxerto Vascular/fisiopatologia , Hemodinâmica , Humanos , Masculino , Pessoa de Meia-Idade , Fragmentos de Peptídeos/farmacologia , Doença Arterial Periférica/diagnóstico , Projetos Piloto , Adesividade Plaquetária/efeitos dos fármacos , Testes de Função Plaquetária , Receptores de Trombina/agonistas , Receptores de Trombina/metabolismo , Medição de Risco , Fatores de Risco , Trombina/metabolismo , Fatores de Tempo , Resultado do Tratamento , Veias/imunologia , Veias/fisiopatologia , Washington
16.
J Biomed Mater Res A ; 95(2): 641-8, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20725965

RESUMO

Based on our discoveries of a unique, synergistic interplay between vascular endothelial growth factor (VEGF) and specific domains of the matrix protein fibronectin (FN), we used recombinant technology to create a new protein construct derived from the cell-binding and VEGF-binding domains of FN. We wished to test the hypothesis that this prototype recombinant FN (rFN) protein would enhance cellular and capillary ingrowth in vivo into expanded polytetrafluoroethylene (ePTFE) implants. ePTFE disks of high porosity (60 micron internodal distance) were embedded with fibrin gel and heparin, with/without mixtures of VEGF and rFN and were implanted subcutaneously in rats. Control implants embedded with fibrin glue and heparin alone showed an average of 8.5% (±0.51% standard error mean (SEM)) cellular ingrowth. The addition of either VEGF or rFN caused a modest but significant increase in cellular ingrowth (12.7 ± 1% and 11.8 ± 0.98%, respectively, p < 0.004). However, the combination of rFN/VEGF/heparin dramatically increased cellular ingrowth (27.6 ± 1.62%, p < 0.001), compared with all other treatments. Quantification of capillary ingrowth yielded the same pattern. These results suggest that the incorporation of such biological modulators into cardiovascular implants could offer new strategies for the design of a ready-made small diameter prosthetic graft with enhanced capacity for neovascularization and endothelialization.


Assuntos
Indutores da Angiogênese/metabolismo , Capilares/fisiologia , Fibronectinas/metabolismo , Implantes Experimentais , Neovascularização Fisiológica , Politetrafluoretileno/química , Proteínas Recombinantes/metabolismo , Animais , Materiais Biocompatíveis/química , Materiais Biocompatíveis/metabolismo , Capilares/citologia , Células Cultivadas , Adesivo Tecidual de Fibrina/metabolismo , Fibronectinas/genética , Heparina/metabolismo , Humanos , Teste de Materiais , Ratos , Ratos Long-Evans , Proteínas Recombinantes/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
17.
J Cell Biochem ; 111(2): 461-8, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20524207

RESUMO

Heparin and heparin-like molecules are known to modulate the cellular responses to vascular endothelial growth factor-A (VEGF-A). In this study, we investigated the likely mechanisms for heparin's influence on the biological activity of VEGF-A. Previous studies have shown that exogenous heparin's effects on the biological activity of VEGF-A are many and varied, in part due to the endogenous cell-surface heparan sulfates. To circumvent this problem, we used mutant endothelial cells lacking cell-surface heparan sulfates. We showed that VEGF-induced cellular responses are dependent in part on the presence of the heparan sulfates, and that exogenous heparin significantly augments VEGF's cellular effects especially when endogenous heparan sulfates are absent. Exogenous heparin was also found to play a cross-bridging role between VEGF-A(165) and putative heparin-binding sites within its cognate receptor, VEGFR2 when they were examined in isolation. The cross-bridging appears to be more dependent on molecular weight than on a specific heparin structure. This was confirmed by surface plasmon resonance binding studies using sugar chips immobilized with defined oligosaccharide structures, which showed that VEGF-A(165) binds to a relatively broad range of sulfated glycosaminoglycan structures. Finally, studies of the far-UV circular dichroism spectra of VEGF-A(165) showed that heparin can also modulate the conformation and secondary structure of the protein.


Assuntos
Células Endoteliais/metabolismo , Heparina/farmacologia , Fator A de Crescimento do Endotélio Vascular/efeitos dos fármacos , Animais , Sítios de Ligação , Glicosaminoglicanos , Heparitina Sulfato , Humanos , Peso Molecular , Conformação Proteica/efeitos dos fármacos , Ressonância de Plasmônio de Superfície , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
18.
Eur J Pharmacol ; 635(1-3): 165-70, 2010 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-20307530

RESUMO

Protamine is the only agent approved to reverse heparin-induced anticoagulation. Due to the significant adverse effects of protamine there is an important need for an alternative agent with an improved safety profile. The pharmacodynamics of PM102, a novel peptide-based heparin antagonist, was evaluated and compared to protamine in a rat model. Rats were dosed with intravenous heparin (50U/kg) and 4min later with protamine (0.25, 0.75mg/kg single intravenous bolus) or PM102 (0.1, 0.3, 1, 3, 30mg/kg single intravenous bolus). Blood samples were collected though 60min for assessment of activated partial thromboplastin time (aPTT) and plasma concentration of PM102. Both doses of protamine markedly lowered the elevated aPTT to baseline values within 1 to 5min after administration. PM102 (0.3-30mg/kg) also rapidly and completely reversed heparin-induced increases in aPTT within 1 to 5min. The effects of PM102 administered as an infusion over 10min also reversed aPTT with similar potency to that observed for bolus administration. The onset of reversal with infusion was delayed relative to the same total dose given as a bolus; however, the maximum effect was similar. PM102 rapidly (T(max) 1-2.6min) appeared in plasma after dosing. Concentrations of PM102 generally declined rapidly after reaching T(max) with a mean T(1/2) of 4 to 31min. PM102 is a novel synthetic peptide that effectively reverses the anticoagulant effect of heparin. It's utility as a bolus injection as well as infusion, its rapid efficacy and its rapid clearance make this an ideal candidate for clinical development.


Assuntos
Antagonistas de Heparina/farmacologia , Heparina/farmacologia , Tempo de Tromboplastina Parcial , Peptídeos/farmacologia , Animais , Anticoagulantes/antagonistas & inibidores , Anticoagulantes/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Relação Dose-Resposta a Droga , Antagonistas de Heparina/administração & dosagem , Antagonistas de Heparina/farmacocinética , Técnicas In Vitro , Injeções Intravenosas , Masculino , Peptídeos/administração & dosagem , Peptídeos/farmacocinética , Protaminas/farmacologia , Ratos , Ratos Sprague-Dawley
19.
Future Cardiol ; 4(4): 409-26, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19804321

RESUMO

Neovacularization is an important biological process whereby new blood vessels develop in both health and disease. During development, blood vessels are formed from mesodermal cells in a process called vasculogenesis. The vascular network then expands by the sprouting of new vessel networks from pre-established vessels in a process known as angiogenesis. However, in adult life, undesirable neovascularization is associated with tumor development and a growing list of 'angiogenesis-dependent' diseases, including cardiovascular complications. Furthermore, diseases characterized by ischemia-induced tissue damage cause a neovascularization response to facilitate tissue repair. Recent research has identified novel molecular and cellular mediators of neovascularization that, in adult life, recapitulate angiogenic processes observed during embryonic development. The discovery of vascular progenitor cells and new molecules that display selective functions in modulating endothelial cell fate, migration and patterning, vessel morphogenesis and the amplification of angiogenic signaling by regulating the master signal VEGF, opens the door to new clinical strategies that target angiogenesis-dependent diseases or that can promote therapeutic neovascularization.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA